Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.610
Filtrar
1.
Front Immunol ; 15: 1405376, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39015565

RESUMEN

Pseudomonas aeruginosa is a highly adaptable opportunistic pathogen capable of exploiting barriers and immune defects to cause chronic lung infections in conditions such as cystic fibrosis. In these contexts, host immune responses are ineffective at clearing persistent bacterial infection, instead driving a cycle of inflammatory lung damage. This review outlines key components of the host immune response to chronic P. aeruginosa infection within the lung, beginning with initial pathogen recognition, followed by a robust yet maladaptive innate immune response, and an ineffective adaptive immune response that propagates lung damage while permitting bacterial persistence. Untangling the interplay between host immunity and chronic P. aeruginosa infection will allow for the development and refinement of strategies to modulate immune-associated lung damage and potentiate the immune system to combat chronic infection more effectively.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Humanos , Pseudomonas aeruginosa/inmunología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Enfermedad Crónica , Animales , Interacciones Huésped-Patógeno/inmunología , Inmunidad Adaptativa , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/microbiología , Fibrosis Quística/inmunología , Fibrosis Quística/microbiología , Fibrosis Quística/complicaciones , Pulmón/inmunología , Pulmón/microbiología
2.
Immunol Lett ; 268: 106886, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38906482

RESUMEN

OBJECTIVE: Novel mRNA-based vaccines have been proven to be powerful tools in combating the global pandemic caused by SARS-CoV-2 protecting individuals, especially the immunocompromised, from COVID-19. Still, it remains largely unknown how solid organ transplant and different immunosuppressive medications affect development of vaccine-induced immunity. METHODS: In this work, we monitored humoral and cellular memory responses after mRNA SARS-CoV-2 two-doses and booster doses vaccination in cystic fibrosis lung transplanted patients (CFT) and compared them with both cystic fibrosis patients without lung transplant (CF) and with kidney transplant recipients (KT). In particular, we investigated the effects of immunosuppressive regimens on immune memory to SARS-CoV-2 after mRNA SARS-CoV-2 vaccine in transplanted patients. RESULTS: Our results showed that immunocompromised transplanted patients displayed a weak cellular and humoral memory to SARS-CoV-2 mRNA vaccination. In addition, obtained data clearly demonstrate that immunosuppressive therapy regimen including antimetabolites, further reduces patients' ability to respond to vaccination at both humoral and cell-mediated level. Notably, patient treated with antimetabolites showed a lower humoral and cellular response also after a booster dose vaccination. CONCLUSION: These results, even if obtained on a small patient's cohort, question whether immunocompromised patients need interventions to improve vaccine SARS-CoV-2 mRNA vaccine response such as additional jab or modulation of immunosuppressive therapy.


Asunto(s)
Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , Inmunidad Celular , Inmunidad Humoral , Huésped Inmunocomprometido , Inmunosupresores , SARS-CoV-2 , Receptores de Trasplantes , Humanos , COVID-19/inmunología , COVID-19/prevención & control , SARS-CoV-2/inmunología , Masculino , Femenino , Inmunosupresores/uso terapéutico , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Adulto , Vacunación , Persona de Mediana Edad , Fibrosis Quística/inmunología , Memoria Inmunológica , Trasplante de Órganos/efectos adversos , Trasplante de Riñón/efectos adversos , Trasplante de Pulmón/efectos adversos , Inmunización Secundaria
3.
Eur J Cell Biol ; 103(2): 151416, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38636185

RESUMEN

Airway epithelial cells form a physical barrier against inhaled pathogens and coordinate innate immune responses in the lungs. Bronchial cells in people with cystic fibrosis (pwCF) are colonized by Pseudomonas aeruginosa because of the accumulation of mucus in the lower airways and an altered immune response. This leads to chronic inflammation, lung tissue damage, and accelerated decline in lung function. Thus, identifying the molecular factors involved in the host response in the airways is crucial for developing new therapeutic strategies. The septin (SEPT) cytoskeleton is involved in tissue barrier integrity and anti-infective responses. SEPT7 is critical for maintaining SEPT complexes and for sensing pathogenic microbes. In the lungs, SEPT7 may be involved in the epithelial barrier resistance to infection; however, its role in cystic fibrosis (CF) P. aeruginosa infection is unknown. This study aimed to investigate the role of SEPT7 in controlling P. aeruginosa infection in bronchial epithelial cells, particularly in CF. The study findings showed that SEPT7 encages P. aeruginosa in bronchial epithelial cells and its inhibition downregulates the expression of other SEPTs. In addition, P. aeruginosa does not regulate SEPT7 expression. Finally, we found that inhibiting SEPT7 expression in bronchial epithelial cells (BEAS-2B 16HBE14o- and primary cells) resulted in higher levels of internalized P. aeruginosa and decreased IL-6 production during infection, suggesting a crucial role of SEPT7 in the host response against this bacterium. However, these effects were not observed in the CF cells (16HBE14o-/F508del and primary cells) which may explain the persistence of infection in pwCF. The study findings suggest the modification of SEPT7 expression as a potential approach for the anti-infective control of P. aeruginosa, particularly in CF.


Asunto(s)
Bronquios , Fibrosis Quística , Células Epiteliales , Pseudomonas aeruginosa , Septinas , Pseudomonas aeruginosa/inmunología , Fibrosis Quística/microbiología , Fibrosis Quística/inmunología , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Humanos , Septinas/metabolismo , Septinas/genética , Células Epiteliales/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/inmunología , Bronquios/microbiología , Bronquios/patología , Bronquios/metabolismo , Bronquios/inmunología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/metabolismo , Línea Celular
4.
PLoS Biol ; 22(4): e3002566, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38652717

RESUMEN

Phage therapy is a therapeutic approach to treat multidrug-resistant (MDR) infections that employs lytic bacteriophages (phages) to eliminate bacteria. Despite the abundant evidence for its success as an antimicrobial in Eastern Europe, there is scarce data regarding its effects on the human host. Here, we aimed to understand how lytic phages interact with cells of the airway epithelium, the tissue site that is colonized by bacterial biofilms in numerous chronic respiratory disorders. Using a panel of Pseudomonas aeruginosa phages and human airway epithelial cells (AECs) derived from a person with cystic fibrosis (CF), we determined that interactions between phages and epithelial cells depend on specific phage properties as well as physiochemical features of the microenvironment. Although poor at internalizing phages, the airway epithelium responds to phage exposure by changing its transcriptional profile and secreting antiviral and proinflammatory cytokines that correlate with specific phage families. Overall, our findings indicate that mammalian responses to phages are heterogenous and could potentially alter the way that respiratory local defenses aid in bacterial clearance during phage therapy. Thus, besides phage receptor specificity in a particular bacterial isolate, the criteria to select lytic phages for therapy should be expanded to include mammalian cell responses.


Asunto(s)
Fibrosis Quística , Citocinas , Células Epiteliales , Pseudomonas aeruginosa , Humanos , Pseudomonas aeruginosa/virología , Células Epiteliales/virología , Células Epiteliales/metabolismo , Células Epiteliales/inmunología , Citocinas/metabolismo , Fibrosis Quística/terapia , Fibrosis Quística/inmunología , Fibrosis Quística/metabolismo , Terapia de Fagos , Bacteriófagos/fisiología , Bacteriófagos/genética , Mucosa Respiratoria/virología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/inmunología , Infecciones por Pseudomonas/terapia , Infecciones por Pseudomonas/inmunología , Fagos Pseudomonas/metabolismo , Biopelículas
5.
Am J Respir Cell Mol Biol ; 71(2): 207-218, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38656811

RESUMEN

Pseudomonas aeruginosa causes chronic lung infection in cystic fibrosis (CF), resulting in structural lung damage and progressive pulmonary decline. P. aeruginosa in the CF lung undergoes numerous changes, adapting to host-specific airway pressures while establishing chronic infection. P. aeruginosa undergoes lipid A structural modification during CF chronic infection that is not seen in any other disease state. Lipid A, the membrane anchor of LPS (i.e., endotoxin), comprises the majority of the outer membrane of Gram-negative bacteria and is a potent Toll-like receptor 4 (TLR4) agonist. The structure of P. aeruginosa lipid A is intimately linked with its recognition by TLR4 and subsequent immune response. Prior work has identified P. aeruginosa strains with altered lipid A structures that arise during chronic CF lung infection; however, the impact of the P. aeruginosa lipid A structure on airway disease has not been investigated. Here, we show that P. aeruginosa lipid A lacks PagL-mediated deacylation during human airway infection using a direct-from-sample mass spectrometry approach on human BAL fluid. This structure triggers increased proinflammatory cytokine production by primary human macrophages. Furthermore, alterations in lipid A 2-hydroxylation impact cytokine response in a site-specific manner, independent of CF transmembrane conductance regulator function. It is interesting that there is a CF-specific reduction in IL-8 secretion within the epithelial-cell compartment that only occurs in CF bronchial epithelial cells when infected with CF-adapted P. aeruginosa that lacks PagL-mediated lipid A deacylation. Taken together, we show that P. aeruginosa alters its lipid A structure during acute lung infection and that this lipid A structure induces stronger signaling through TLR4.


Asunto(s)
Fibrosis Quística , Lípido A , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/inmunología , Humanos , Lípido A/metabolismo , Lípido A/inmunología , Fibrosis Quística/microbiología , Fibrosis Quística/inmunología , Fibrosis Quística/metabolismo , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/inmunología , Citocinas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Líquido del Lavado Bronquioalveolar/inmunología , Pulmón/microbiología , Pulmón/inmunología , Pulmón/metabolismo
6.
Tuberculosis (Edinb) ; 147: 102504, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38522174

RESUMEN

Mycobacterium tuberculosis and opportunistic environmental non-tuberculous mycobacteria (NTM) can cause severe infection. Why latent tuberculosis infection advances to active disease, and why some individuals with cystic fibrosis (CF) develop pulmonary infections with NTM is still poorly understood. The aim of this study was to investigate the effector function of peripheral blood mononuclear cells (PBMC) from individuals with active or latent tuberculosis, individuals with CF with or without pulmonary NTM-infection and healthy controls, by measuring cytokine response to in vitro stimulation with different species of NTMs. The cytokine concentrations of IL-17A, IL-22, IL-23, IL-10, IL12p70 and IFN-γ were measured in PBMC-culture supernatants after stimulation with NTMs. PBMCs from individuals with latent tuberculosis infection showed strong IL-17A, IL-22, and IFN-γ responses compared to individuals with active tuberculosis or CF. IL-10 production was low in both tuberculosis groups compared to the CF groups and controls. This study suggests that IL-17A and IL-22 might be important to keep tuberculosis in a latent phase and that individuals with CF with an ongoing NTM infection seem to have a low cytokine response.


Asunto(s)
Fibrosis Quística , Citocinas , Tuberculosis Latente , Leucocitos Mononucleares , Infecciones por Mycobacterium no Tuberculosas , Micobacterias no Tuberculosas , Humanos , Fibrosis Quística/microbiología , Fibrosis Quística/inmunología , Tuberculosis Latente/inmunología , Tuberculosis Latente/microbiología , Femenino , Masculino , Adulto , Micobacterias no Tuberculosas/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/microbiología , Citocinas/metabolismo , Estudios de Casos y Controles , Infecciones por Mycobacterium no Tuberculosas/inmunología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Células Cultivadas , Persona de Mediana Edad , Adulto Joven , Interleucinas/metabolismo , Interleucinas/sangre , Interleucinas/inmunología , Interferón gamma/metabolismo , Interferón gamma/inmunología , Interleucina-17/metabolismo , Interleucina-22 , Adolescente , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/sangre
8.
Am J Respir Crit Care Med ; 209(11): 1338-1350, 2024 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-38259174

RESUMEN

Rationale: Pharmacological improvement of cystic fibrosis transmembrane conductance regulator (CFTR) function with elexacaftor/tezacaftor/ivacaftor (ETI) provides unprecedented improvements in lung function and other clinical outcomes in patients with cystic fibrosis (CF). However, ETI effects on impaired mucosal homeostasis and host defense at the molecular and cellular levels in the airways of patients with CF remain unknown. Objectives: To investigate effects of ETI on the transcriptome of nasal epithelial and immune cells from children with CF at the single-cell level. Methods: Nasal swabs from 13 children with CF and at least one F508del allele aged 6 to 11 years were collected at baseline and 3 months after initiation of ETI, subjected to single-cell RNA sequencing, and compared with swabs from 12 age-matched healthy children. Measurements and Main Results: Proportions of CFTR-positive cells were decreased in epithelial basal, club, and goblet cells, but not in ionocytes, from children with CF at baseline and were restored by ETI therapy to nearly healthy levels. Single-cell transcriptomics revealed an impaired IFN signaling and reduced expression of major histocompatibility complex classes I and II encoding genes in epithelial cells of children with CF at baseline, which was partially restored by ETI. In addition, ETI therapy markedly reduced the inflammatory phenotype of immune cells, particularly of neutrophils and macrophages. Conclusions: Pharmacological improvement of CFTR function improves innate mucosal immunity and reduces immune cell inflammatory responses in the upper airways of children with CF at the single-cell level, highlighting the potential to restore epithelial homeostasis and host defense in CF airways by early initiation of ETI therapy.


Asunto(s)
Aminofenoles , Benzodioxoles , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Homeostasis , Humanos , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/inmunología , Fibrosis Quística/fisiopatología , Niño , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Femenino , Masculino , Benzodioxoles/uso terapéutico , Benzodioxoles/farmacología , Aminofenoles/uso terapéutico , Aminofenoles/farmacología , Quinolonas/uso terapéutico , Quinolonas/farmacología , Indoles/uso terapéutico , Indoles/farmacología , Combinación de Medicamentos , Quinolinas/uso terapéutico , Quinolinas/farmacología , Pirazoles/uso terapéutico , Pirazoles/farmacología , Pirroles/uso terapéutico , Pirroles/farmacología , Mucosa Nasal/inmunología , Piridinas/uso terapéutico , Piridinas/farmacología
9.
Front Biosci (Landmark Ed) ; 27(5): 149, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35638416

RESUMEN

BACKGROUND: People with Cystic Fibrosis (CF) develop pulmonary inflammation, chronic infection and structural lung damage early in life, with these manifestations being prevalent among preschool children and infants. While early immune events are believed to play critical roles in shaping the progression, severity and disease burden later in life, T cells and their subsets are poorly studied in the CF lung, particularly during the formative early stages of disease. METHODS: Using flow cytometry, we analyzed Mucosal Associated Invariant T (MAIT) cells, γδ T cells, and Natural Killer T (NKT)-like cells in bronchoalveolar lavage (BAL) samples from seventeen children with CF, aged two to six years old. The effect of age, sex and lung infections on the frequencies of these cells in BAL samples was analysed (grouped data were tested for normality and compared by t-test or Kruskal-Wallis analysis). RESULTS: No difference was noted in the proportions of unconventional T cells related to the sex or age of the children. The frequency of γδ T cells and MAIT cells appeared unchanged by infection status. However, viral infections were associated with a significant increase in the proportion of NKT-like cells. CONCLUSIONS: By evaluating T cells in the lungs of children during the early formative stages of CF, this study identified potentially important interactions between these cells and viral pathogens.


Asunto(s)
Fibrosis Quística , Linfocitos T/inmunología , Virosis , Niño , Preescolar , Fibrosis Quística/complicaciones , Fibrosis Quística/inmunología , Fibrosis Quística/virología , Humanos , Lactante , Pulmón/inmunología , Pulmón/virología
10.
JCI Insight ; 7(6)2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35315363

RESUMEN

Cystic fibrosis (CF) is an inherited disorder caused by biallelic mutations of the CF transmembrane conductance regulator (CFTR) gene. Converging evidence suggests that CF carriers with only 1 defective CFTR copy are at increased risk for CF-related conditions and pulmonary infections, but the molecular mechanisms underpinning this effect remain unknown. We performed transcriptomic profiling of peripheral blood mononuclear cells (PBMCs) of CF child-parent trios (proband, father, and mother) and healthy control (HC) PBMCs or THP-1 cells incubated with the plasma of these participants. Transcriptomic analyses revealed suppression of cytokine-enriched immune-related genes (IL-1ß, CXCL8, CREM), implicating lipopolysaccharide tolerance in innate immune cells (monocytes) of CF probands and their parents. These data suggest that a homozygous as well as a heterozygous CFTR mutation can modulate the immune/inflammatory system. This conclusion is further supported by the finding of lower numbers of circulating monocytes in CF probands and their parents, compared with HCs, and the abundance of mononuclear phagocyte subsets, which correlated with Pseudomonas aeruginosa infection, lung disease severity, and CF progression in the probands. This study provides insight into demonstrated CFTR-related innate immune dysfunction in individuals with CF and carriers of a CFTR mutation that may serve as a target for personalized therapy.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Macrófagos , Monocitos , Fibrosis Quística/genética , Fibrosis Quística/inmunología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Leucocitos Mononucleares , Macrófagos/patología , Monocitos/patología , Padres
11.
J Immunol ; 208(4): 979-990, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35046105

RESUMEN

Calprotectin is released by activated neutrophils along with myeloperoxidase (MPO) and proteases. It plays numerous roles in inflammation and infection, and is used as an inflammatory biomarker. However, calprotectin is readily oxidized by MPO-derived hypohalous acids to form covalent dimers of its S100A8 and S100A9 subunits. The dimers are susceptible to degradation by proteases. We show that detection of human calprotectin by ELISA declines markedly because of its oxidation by hypochlorous acid and subsequent degradation. Also, proteolysis liberates specific peptides from oxidized calprotectin that is present at inflammatory sites. We identified six calprotectin-derived peptides by mass spectrometry and detected them in the bronchoalveolar lavage fluid of children with cystic fibrosis (CF). We assessed the peptides as biomarkers of neutrophilic inflammation and infection. The content of the calprotectin peptide ILVI was related to calprotectin (r = 0.72, p = 0.01, n = 10). Four of the peptides were correlated with the concentration of MPO (r > 0.7, p ≤ 0.01, n = 21), while three were higher (p < 0.05) in neutrophil elastase-positive (n = 14) than -negative samples (n = 7). Also, five of the peptides were higher (p < 0.05) in the bronchoalveolar lavage fluid from children with CF with infections (n = 21) than from non-CF children without infections (n = 6). The specific peptides liberated from calprotectin will signal uncontrolled activity of proteases and MPO during inflammation. They may prove useful in tracking inflammation in respiratory diseases dominated by neutrophils, including coronavirus disease 2019.


Asunto(s)
Líquido del Lavado Bronquioalveolar/inmunología , Fibrosis Quística/inmunología , Inflamación/inmunología , Complejo de Antígeno L1 de Leucocito/metabolismo , Neutrófilos/inmunología , Péptidos/metabolismo , Sistema Respiratorio/metabolismo , Niño , Preescolar , Fibrosis Quística/diagnóstico , Femenino , Humanos , Inflamación/diagnóstico , Complejo de Antígeno L1 de Leucocito/genética , Complejo de Antígeno L1 de Leucocito/inmunología , Masculino , Activación Neutrófila , Oxidación-Reducción , Péptidos/genética , Péptidos/inmunología , Proteolisis
12.
Microbiol Spectr ; 10(1): e0254621, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35080463

RESUMEN

Mycobacterium abscessus is the etiological agent of severe pulmonary infections in vulnerable patients, such as those with cystic fibrosis (CF), where it represents a relevant cause of morbidity and mortality. Treatment of pulmonary infections caused by M. abscessus remains extremely difficult, as this species is resistant to most classes of antibiotics, including macrolides, aminoglycosides, rifamycins, tetracyclines, and ß-lactams. Here, we show that apoptotic body like liposomes loaded with phosphatidylinositol 5-phosphate (ABL/PI5P) enhance the antimycobacterial response, both in macrophages from healthy donors exposed to pharmacological inhibition of cystic fibrosis transmembrane conductance regulator (CFTR) and in macrophages from CF patients, by enhancing phagosome acidification and reactive oxygen species (ROS) production. The treatment with liposomes of wild-type as well as CF mice, intratracheally infected with M. abscessus, resulted in about a 2-log reduction of pulmonary mycobacterial burden and a significant reduction of macrophages and neutrophils in bronchoalveolar lavage fluid (BALF). Finally, the combination treatment with ABL/PI5P and amikacin, to specifically target intracellular and extracellular bacilli, resulted in a further significant reduction of both pulmonary mycobacterial burden and inflammatory response in comparison with the single treatments. These results offer the conceptual basis for a novel therapeutic regimen based on antibiotic and bioactive liposomes, used as a combined host- and pathogen-directed therapeutic strategy, aimed at the control of M. abscessus infection, and of related immunopathogenic responses, for which therapeutic options are still limited. IMPORTANCE Mycobacterium abscessus is an opportunistic pathogen intrinsically resistant to many antibiotics, frequently linked to chronic pulmonary infections, and representing a relevant cause of morbidity and mortality, especially in immunocompromised patients, such as those affected by cystic fibrosis. M. abscessus-caused pulmonary infection treatment is extremely difficult due to its high toxicity and long-lasting regimen with life-impairing side effects and the scarce availability of new antibiotics approved for human use. In this context, there is an urgent need for the development of an alternative therapeutic strategy that aims at improving the current management of patients affected by chronic M. abscessus infections. Our data support the therapeutic value of a combined host- and pathogen-directed therapy as a promising approach, as an alternative to single treatments, to simultaneously target intracellular and extracellular pathogens and improve the clinical management of patients infected with multidrug-resistant pathogens such as M. abscessus.


Asunto(s)
Antibacterianos/administración & dosificación , Fibrosis Quística/inmunología , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Mycobacterium abscessus/efectos de los fármacos , Fosfatos de Fosfatidilinositol/administración & dosificación , Amicacina/administración & dosificación , Amicacina/química , Animales , Antibacterianos/química , Fibrosis Quística/complicaciones , Fibrosis Quística/genética , Fibrosis Quística/microbiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/inmunología , Femenino , Humanos , Liposomas/química , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Mycobacterium no Tuberculosas/etiología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium abscessus/fisiología , Fagosomas/inmunología , Fosfatos de Fosfatidilinositol/química , Especies Reactivas de Oxígeno/inmunología
13.
Cells ; 10(12)2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34944110

RESUMEN

The respiratory epithelium represents the first chemical, immune, and physical barrier against inhaled noxious materials, particularly pathogens in cystic fibrosis. Local mucus thickening, altered mucociliary clearance, and reduced pH due to CFTR protein dysfunction favor bacterial overgrowth and excessive inflammation. We aimed in this review to summarize respiratory mucosal alterations within the epithelium and current knowledge on local immunity linked to immunoglobulin A in patients with cystic fibrosis.


Asunto(s)
Fibrosis Quística/inmunología , Inmunidad Mucosa/inmunología , Inmunoglobulina A/metabolismo , Mucosa Respiratoria/inmunología , Animales , Fibrosis Quística/patología , Humanos , Pulmón/patología , Modelos Biológicos
14.
Front Immunol ; 12: 733217, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721395

RESUMEN

The immune landscape of the paediatric respiratory system remains largely uncharacterised and as a result, the mechanisms of globally important childhood respiratory diseases remain poorly understood. In this work, we used high parameter flow cytometry and inflammatory cytokine profiling to map the local [bronchoalveolar lavage (BAL)] and systemic (whole blood) immune response in preschool aged children with cystic fibrosis (CF) and aged-matched healthy controls. We demonstrate that children with CF show pulmonary infiltration of CD66b+ granulocytes and increased levels of MIP-1α, MIG, MCP-1, IL-8, and IL-6 in BAL relative to healthy control children. Proportions of systemic neutrophils positively correlated with age in children with CF, whilst systemic CD4 T cells and B cells were inversely associated with age. Inflammatory cells in the BAL from both CF and healthy children expressed higher levels of activation and migration markers relative to their systemic counterparts. This work highlights the utility of multiplex immune profiling and advanced analytical pipelines to understand mechanisms of lung disease in childhood.


Asunto(s)
Fibrosis Quística/inmunología , Citocinas/sangre , Mediadores de Inflamación/sangre , Inflamación/inmunología , Leucocitos/inmunología , Neumonía/inmunología , Biomarcadores/sangre , Líquido del Lavado Bronquioalveolar/inmunología , Estudios de Casos y Controles , Fibrosis Quística/diagnóstico , Fibrosis Quística/metabolismo , Citometría de Flujo , Humanos , Inmunofenotipificación , Inflamación/diagnóstico , Inflamación/metabolismo , Leucocitos/metabolismo , Fenotipo , Neumonía/diagnóstico , Neumonía/metabolismo
15.
Front Immunol ; 12: 745326, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34621276

RESUMEN

Cystic Fibrosis (CF) is a genetic disease that causes chronic and severe lung inflammation and infection associated with high rates of mortality. In CF, disrupted ion exchange in the epithelium results in excessive mucus production and reduced mucociliary clearance, leading to immune system exacerbation and chronic infections with pathogens such as P. aeruginosa and S. aureus. Constant immune stimulation leads to altered immune responses including T cell impairment and neutrophil dysfunction. Specifically, CF is considered a Th17-mediated disease, and it has been proposed that both P. aeruginosa and a subset of neutrophils known as granulocytic myeloid suppressor cells (gMDSCs) play a role in T cell suppression. The exact mechanisms behind these interactions are yet to be determined, but recent works demonstrate a role for arginase-1. It is also believed that P. aeruginosa drives gMDSC function as a means of immune evasion, leading to chronic infection. Herein, we review the current literature regarding immune suppression in CF by gMDSCs with an emphasis on T cell impairment and the role of P. aeruginosa in this dynamic interaction.


Asunto(s)
Fibrosis Quística/inmunología , Granulocitos/inmunología , Evasión Inmune , Células Supresoras de Origen Mieloide/inmunología , Pseudomonas aeruginosa/inmunología , Células Th17/inmunología , Arginasa/fisiología , Fibrosis Quística/complicaciones , Citotoxicidad Inmunológica , Humanos , Neutrófilos/inmunología , Neutrófilos/patología , Infección Persistente , Infecciones por Pseudomonas/complicaciones , Infecciones por Pseudomonas/inmunología , Linfocitos T Reguladores/inmunología
16.
Sci Rep ; 11(1): 20082, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34635725

RESUMEN

Invariant Natural Killer T (iNKT) cells respond to the ligation of lipid antigen-CD1d complexes via their T-cell receptor and are implicated in various immunometabolic diseases. We considered that immunometabolic factors might affect iNKT cell function. To this end, we investigated iNKT cell phenotype and function in a cohort of adolescents with chronic disease and immunometabolic abnormalities. We analyzed peripheral blood iNKT cells of adolescents with cystic fibrosis (CF, n = 24), corrected coarctation of the aorta (CoA, n = 25), juvenile idiopathic arthritis (JIA, n = 20), obesity (OB, n = 20), and corrected atrial septal defect (ASD, n = 25) as controls. To study transcriptional differences, we performed RNA sequencing on a subset of obese patients and controls. Finally, we performed standardized co-culture experiments using patient plasma, to investigate the effect of plasma factors on iNKT cell function. We found comparable iNKT cell numbers across patient groups, except for reduced iNKT cell numbers in JIA patients. Upon ex-vivo activation, we observed enhanced IFN-γ/IL-4 cytokine ratios in iNKT cells of obese adolescents versus controls. The Th1-skewed iNKT cell cytokine profile of obese adolescents was not explained by a distinct transcriptional profile of the iNKT cells. Co-culture experiments with patient plasma revealed that across all patient groups, obesity-associated plasma factors including LDL-cholesterol, leptin, and fatty-acid binding protein 4 (FABP4) coincided with higher IFN-γ production, whereas high HDL-cholesterol and insulin sensitivity (QUICKI) coincided with higher IL-4 production. LDL and HDL supplementation in co-culture studies confirmed the effects of lipoproteins on iNKT cell cytokine production. These results suggest that circulating immunometabolic factors such as lipoproteins may be involved in Th1 skewing of the iNKT cell cytokine response in immunometabolic disease.


Asunto(s)
Artritis Juvenil/inmunología , Fibrosis Quística/inmunología , Defectos del Tabique Interatrial/inmunología , Células T Asesinas Naturales/inmunología , Obesidad/fisiopatología , Células TH1/inmunología , Adolescente , Artritis Juvenil/metabolismo , Artritis Juvenil/patología , Estudios de Casos y Controles , Enfermedad Crónica , Estudios Transversales , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Citocinas/metabolismo , Femenino , Defectos del Tabique Interatrial/metabolismo , Defectos del Tabique Interatrial/patología , Humanos , Interferón gamma/metabolismo , Masculino
17.
J Immunol ; 207(4): 1165-1179, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34330754

RESUMEN

Increased levels of ambient ozone, one of the six criteria air pollutants, result in respiratory tract injury and worsening of ongoing lung diseases. However, the effect of ozone exposure on the respiratory tract undergoing active lung development and simultaneously experiencing mucoinflammatory lung diseases, such as cystic fibrosis, remains unclear. To address these questions, we exposed Scnn1b transgenic (Scnn1b-Tg+) mice, a mouse model of cystic fibrosis-like lung disease, and littermate wild-type (WT) mice to ozone from postnatal days (PND) 3-20 and examined the lung phenotypes at PND21. As compared with filtered air (FA)-exposed WT mice, the ozone-exposed WT mice exhibited marked alveolar space enlargement, in addition to significant eosinophilic infiltration, type 2 inflammation, and mucous cell metaplasia. Ozone-exposed Scnn1b-Tg+ mice also exhibited significantly increased alveolar space enlargement, which was also accompanied by exaggerated granulocytic infiltration, type 2 inflammation, and a greater degree of mucus obstruction. The alveolar space enlargement in ozone-exposed WT, FA-exposed Scnn1b-Tg+, and ozone-exposed Scnn1b-Tg+ mice was accompanied by elevated levels of MMP12 protein in macrophages and Mmp12 mRNA in the lung homogenates. Finally, although bacterial burden was largely resolved by PND21 in FA-exposed Scnn1b-Tg+ mice, ozone-exposed Scnn1b-Tg+ mice exhibited compromised bacterial clearance, which was also associated with increased levels of IL-10, an immunosuppressive cytokine, and marked mucus obstruction. Taken together, our data show that ozone exposure results in alveolar space remodeling during active phases of lung development and markedly exaggerates the mucoinflammatory outcomes of pediatric-onset lung disease, including bacterial infections, granulocytic inflammation, mucus obstruction, and alveolar space enlargement.


Asunto(s)
Bacterias/inmunología , Canales Epiteliales de Sodio/inmunología , Inflamación/inmunología , Pulmón/inmunología , Ozono/efectos adversos , Animales , Fibrosis Quística/inmunología , Modelos Animales de Enfermedad , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Moco/inmunología , Atención Posnatal
18.
Front Immunol ; 12: 625922, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34168640

RESUMEN

Low density neutrophils (LDNs) are described in a number of inflammatory conditions, cancers and infections and associated with immunopathology, and a mechanistic role in disease. The role of LDNs at homeostasis in healthy individuals has not been investigated. We have developed an isolation protocol that generates high purity LDNs from healthy donors. Healthy LDNs were identical to healthy normal density neutrophils (NDNs), aside from reduced neutrophil extracellular trap formation. CD66b, CD16, CD15, CD10, CD54, CD62L, CXCR2, CD47 and CD11b were expressed at equivalent levels in healthy LDNs and NDNs and underwent apoptosis and ROS production interchangeably. Healthy LDNs had no differential effect on CD4+ or CD8+ T cell proliferation or IFNγ production compared with NDNs. LDNs were generated from healthy NDNs in vitro by activation with TNF, LPS or fMLF, suggesting a mechanism of LDN generation in disease however, we show neutrophilia in people with Cystic Fibrosis (CF) was not due to increased LDNs. LDNs are present in the neutrophil pool at homeostasis and have limited functional differences to NDNs. We conclude that increased LDN numbers in disease reflect the specific pathology or inflammatory environment and that neutrophil density alone is inadequate to classify discrete functional populations of neutrophils.


Asunto(s)
Separación Celular , Citometría de Flujo , Neutrófilos/inmunología , Antígenos CD/metabolismo , Apoptosis , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Microambiente Celular , Centrifugación por Gradiente de Densidad , Fibrosis Quística/inmunología , Fibrosis Quística/metabolismo , Trampas Extracelulares/metabolismo , Voluntarios Sanos , Homeostasis , Humanos , Interferón gamma/metabolismo , Recuento de Leucocitos , Trastornos Leucocíticos/inmunología , Trastornos Leucocíticos/metabolismo , Activación de Linfocitos , Activación Neutrófila , Neutrófilos/metabolismo , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-8B/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
19.
mSphere ; 6(3): e0035821, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34160233

RESUMEN

Staphylococcus aureus is one of the most common pathogens isolated from the airways of cystic fibrosis (CF) patients and often persists for extended periods. There is limited knowledge about the diversity of S. aureus in CF. We hypothesized that increased diversity of S. aureus would impact CF lung disease. Therefore, we conducted a 1-year observational prospective study with 14 patients with long-term S. aureus infection. From every sputum, 40 S. aureus isolates were chosen and characterized in terms of phenotypic appearance (size, hemolysis, mucoidy, and pigmentation), important virulence traits such as nuclease activity, biofilm formation, and molecular typing by spa sequence typing. Data about coinfection with Pseudomonas aeruginosa and clinical parameters such as lung function, exacerbation, and inflammatory markers in blood (C-reactive protein [CRP], interleukin 6 [IL-6], and S100A8/9 [calprotectin]) were collected. From 58 visits of 14 patients, 2,319 S. aureus isolates were distinguished into 32 phenotypes (PTs) and 50 spa types. The Simpson diversity index (SDI) was used to calculate the phenotypic and genotypic diversity, revealing a high diversity of PTs ranging from 0.19 to 0.87 among patients, while the diversity of spa types of isolates was less pronounced. The SDI of PTs was positively associated with P. aeruginosa coinfection and inflammatory parameters, with IL-6 being the most sensitive parameter. Also, coinfection with P. aeruginosa was associated with mucoid S. aureus and S. aureus with high nuclease activity. Our analyses showed that in CF patients with long-term S. aureus airway infection, a highly diverse and dynamic S. aureus population was present and associated with P. aeruginosa coinfection and inflammation. IMPORTANCE Staphylococcus aureus can persist for extended periods in the airways of people with cystic fibrosis (CF) in spite of antibiotic therapy and high numbers of neutrophils, which fail to eradicate this pathogen. Therefore, S. aureus needs to adapt to this hostile niche. There is only limited knowledge about the diversity of S. aureus in respiratory specimens. We conducted a 1-year prospective study with 14 patients with long-term S. aureus infection and investigated 40 S. aureus isolates from every sputum in terms of phenotypic appearance, nuclease activity, biofilm formation, and molecular typing. Data about coinfection with Pseudomonas aeruginosa and clinical parameters such as lung function, exacerbation, and inflammatory markers in blood were collected. Thirty-two phenotypes (PTs) and 50 spa types were distinguished. Our analyses revealed that in CF patients with long-term S. aureus airway infection, a highly diverse and dynamic S. aureus population was associated with P. aeruginosa coinfection and inflammation.


Asunto(s)
Coinfección/inmunología , Fibrosis Quística/microbiología , Inflamación/microbiología , Infecciones por Pseudomonas/inmunología , Enfermedades Respiratorias/microbiología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/genética , Adaptación Fisiológica , Adolescente , Adulto , Biopelículas/crecimiento & desarrollo , Fibrosis Quística/inmunología , Femenino , Genotipo , Humanos , Inflamación/inmunología , Masculino , Persona de Mediana Edad , Fenotipo , Estudios Prospectivos , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/patogenicidad , Esputo/microbiología , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Virulencia , Adulto Joven
20.
J Clin Invest ; 131(16)2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34166230

RESUMEN

Without cystic fibrosis transmembrane conductance regulator-mediated (CFTR-mediated) HCO3- secretion, airway epithelia of newborns with cystic fibrosis (CF) produce an abnormally acidic airway surface liquid (ASL), and the decreased pH impairs respiratory host defenses. However, within a few months of birth, ASL pH increases to match that in non-CF airways. Although the physiological basis for the increase is unknown, this time course matches the development of inflammation in CF airways. To learn whether inflammation alters CF ASL pH, we treated CF epithelia with TNF-α and IL-17 (TNF-α+IL-17), 2 inflammatory cytokines that are elevated in CF airways. TNF-α+IL-17 markedly increased ASL pH by upregulating pendrin, an apical Cl-/HCO3- exchanger. Moreover, when CF epithelia were exposed to TNF-α+IL-17, clinically approved CFTR modulators further alkalinized ASL pH. As predicted by these results, in vivo data revealed a positive correlation between airway inflammation and CFTR modulator-induced improvement in lung function. These findings suggest that inflammation is a key regulator of HCO3- secretion in CF airways. Thus, they explain earlier observations that ASL pH increases after birth and indicate that, for similar levels of inflammation, the pH of CF ASL is abnormally acidic. These results also suggest that a non-cell-autonomous mechanism, airway inflammation, is an important determinant of the response to CFTR modulators.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Interleucina-17/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Aminofenoles/administración & dosificación , Benzodioxoles/administración & dosificación , Bicarbonatos/metabolismo , Células Cultivadas , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/inmunología , Fibrosis Quística/fisiopatología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos de los fármacos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Combinación de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Indoles/administración & dosificación , Lactante , Recién Nacido , Interleucina-17/administración & dosificación , Transporte Iónico , Mutación , Pirazoles/administración & dosificación , Piridinas/administración & dosificación , Quinolinas/administración & dosificación , Mucosa Respiratoria/efectos de los fármacos , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo , Factor de Necrosis Tumoral alfa/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA